Lgr5-Mediated Restraint of Β-Catenin Is Essential for B-Lymphopoiesis and Leukemia-Initiation

Total Page:16

File Type:pdf, Size:1020Kb

Lgr5-Mediated Restraint of Β-Catenin Is Essential for B-Lymphopoiesis and Leukemia-Initiation bioRxiv preprint doi: https://doi.org/10.1101/2020.03.12.989277; this version posted March 13, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Letter February 5, 2020 2020-02-01971B Lgr5-mediated restraint of β-catenin is essential for B-lymphopoiesis and leukemia-initiation Kadriye Nehir Cosgun1, Mark E. Robinson1, Gauri Deb1, Xin Yang2, Gang Xiao1, Teresa Sadras1, Jaewoong Lee1, Lai N. Chan1, Kohei Kume1, Maurizio Mangolini3, Janet Winchester1, Zhengshan Chen1, Lu Yang1, Huimin Geng2, Shai Izraeli1,4, Joo Song5, Wing-Chung Chan5, Andrew G. Polson6, Hassan Jumaa7, Hans Clevers8 & Markus Müschen1 1Department of Systems Biology, City of Hope Comprehensive Cancer Center, Monrovia, CA 91016 2Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA 94143 3Department of Haematology, Cambridge University, Cambridge, UK 4 Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel 5Department of Pathology, City of Hope Comprehensive Cancer Center, Duarte, CA 6Genentech, Inc., South San Francisco, CA 94080 7Institute for Immunology, University of Ulm, Ulm, Germany 89081 8Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Centre Utrecht, Utrecht, Netherlands For correspondence: Markus Müschen, MD-PhD E-mail: [email protected] Phone: +1-626-218-5171 Department of Systems Biology, City of Hope Comprehensive Cancer Center, 1218 South Fifth Ave, Monrovia, CA 91016 bioRxiv preprint doi: https://doi.org/10.1101/2020.03.12.989277; this version posted March 13, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Upon productive immunoglobulin gene rearrangement, expression of a functional pre-B cell receptor (pre-BCR) initiates positive selection of pre-B cells, clonal expansion and self-renewal1-2. Studying mechanisms driving this first wave of B-lymphopoiesis, we identified the G-protein coupled receptor Lgr5 as an essential initiator of positive selection. Lgr5 was extensively studied as determinant of stem cell populations in multiple tissues3-6, but not in B-cells. While undetectable throughout the hematopoietic system, positively selected pre-B cells were marked with a sharp peak of Lgr5 expression. Conditional deletion of Lgr5 preceding the pre-BCR checkpoint induced negative selection and complete abortion of B-cell development. Proteomic studies of Lgr5-ablation revealed massive (>250-fold) accumulation of -catenin and suppression of MYC. Lgr5-deficient pre-B cells fully recovered by concurrent -catenin-deletion, demonstrating a central role of Lgr5-mediated restraint of -catenin at the pre-BCR checkpoint. In other cell types, -catenin/TCF4 complexes drive transcriptional activation of MYC7-9. Instead of TCF4, proximity-based interactome studies in pre-B cells identified the B- lymphoid transcription factors IKZF1 and IKZF310-11 as -catenin-binding partners, which had the opposite effect and caused transcriptional repression of MYC. On positively selected pre-B cells, Lgr5 prevented accumulation of -catenin and formation of complexes with IKZF1 and IKZF3, which relieved transcriptional repression of MYC. Activating β-catenin-mutations are common throughout all main types of cancer7-8, but were conspicuously absent in pre-B leukemia (B-ALL). Like pre-B cells, B- ALL cells were uniquely sensitive to genetic and pharmacological β-catenin hyperactivation, which recapitulated the effects of Lgr5-deletion and compromised colony formation and leukemia-initiation. A new LGR5 antibody-drug conjugate targeted leukemia-initiating cells in patient-derived B-ALL and achieved long-term disease-control. Likewise, small molecule hyperactivation of -catenin selectively killed B-ALL but not other cell types. Hence, Lgr5-mediated restraint of -catenin activation is essential for B-lymphopoiesis and revealed an unexpected vulnerability that can be leveraged for the treatment of drug-resistant B-ALL. Lgr5 is essential for the initiation of B-lymphopoiesis Once early B-lymphocyte precursors have productively rearranged immunoglobulin (Ig) V, D and J gene segments, expression of a functional Ig -heavy chain as part of the pre-B cell receptor (pre-BCR) results in a strong positive selection signal to initiate clonal expansion and the first wave of B-lymphopoiesis1, 2. To study mechanisms of positive selection and pre-B cell self-renewal, we analyzed gene expression changes that are induced at the onset of Ig -heavy chain signaling. Among the most prominent changes at the pre-BCR checkpoint, we identified upregulation of the Leucine-rich repeat-containing G-protein coupled receptor 5 (Lgr5; Fig. 1a and b, Extended data figure 1a). While mRNA levels of Lgr5 were low or undetectable 2 bioRxiv preprint doi: https://doi.org/10.1101/2020.03.12.989277; this version posted March 13, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. throughout the entire spectrum of hematopoietic lineages, the pre-BCR checkpoint was marked by a sharp peak of Lgr5 expression (Hardy Fractions C-D; Fig. 1c, Extended data figure 1b-c). Lgr5 was extensively studied as determinant of adult stem cell populations in multiple tissues including intestinal3, kidney, liver4 and gastric tissues as well as ear and hair follicles5. For instance, in the colon crypt, Lgr5 marks quiescent stem cell populations that promote tissue regeneration and give rise to the main lineages of the colon epithelium6. Mechanistically, Lgr5 is thought to interact with Rspo1 to potentiate WNT signaling by stabilizing Frizzled (FZD) surface receptors of WNT12, induce assembly of disheveled (DVL2)13-16 and -arrestin (ARRB2)17-20 scaffolds, which ultimately results in accumulation of -catenin. A role of Lgr5 was not previously examined in B-lymphocyte development. To study the functional significance of sharp upregulation of Lgr5 at the pre-BCR checkpoint (Hardy Fractions C-D), we tested the consequences of inducible Cre-mediated deletion of Lgr5 in developing pre-B cells. Under cell culture conditions, inducible Lgr5-deletion caused rapid loss of competitive fitness of pre-B cells (Fig. 1d). In a conditional mouse model for deletion of Lgr5 from earliest stages of B-cell development (Mb1-Cre), pro-B cells (Hardy Fractions A-B) developed normally. However, B cell precursors could not progress past the pre- BCR checkpoint resulting in complete abortion of B-cell development in these mice (Fig. 1e-f, Extended data figure 2). Even deletion of only one allele of Lgr5 was sufficient to cause near-complete ablation of B-cell development, suggesting that even moderate reduction of Lgr5 gene expression may compromise positive selection at the pre-BCR checkpoint (Fig. 1e-f, Extended data figure 2). Interestingly, while deletion of Lgr5 compromised B-lymphopoiesis in vivo and competitive fitness in vitro, loss of Lgr5 did not significantly increase apoptosis and cell death (Extended data figure 9d, f). In addition, when deletion of Lgr5 was induced in mature B-cells after the pre-BCR checkpoint (Cd21-Cre; Extended data figure 3) or in germinal center B- cells (Aicda-Cre; Extended data figure 4), loss of Lgr5 only caused a modest reduction of marginal zone B- cells. All other mature B-cell subsets, including B1 cells remained unaffected. Lgr5-deficient germinal center (GC) B-cells underwent normal affinity maturation and were able to develop antigen-specific B cells in response to immunization (Extended data figure 4). Consistent with a sharp peak of gene expression in pre- B cells (Hardy Fractions C-D), these findings indicate that Lgr5 is uniquely required for positive selection at the pre-BCR checkpoint. Once mature B-cell populations have formed, they no longer depend on Lgr5 function for maintenance and affinity maturation in humoral immune responses. Lgr5 expression predicts poor clinical outcomes in patients with B-ALL While Lgr5 contributes to self-renewal of normal epithelial stem cells and tissue regeneration, this is also the case for cancer-initiating cells. For instance, lineage-tracing experiments revealed Lgr5 as determinant of tumor-initiation in intestinal adenomas21. In addition, Lgr5 marks colon cancer-initiating cells22, promotes metastasis23 and represents a therapeutic target for eradication of colon cancer stem cells (NCT02726334, 3 bioRxiv preprint doi: https://doi.org/10.1101/2020.03.12.989277; this version posted March 13, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. NCT03526835)24, 25. For this reason, we studied LGR5 mRNA levels in patient-derived samples of B-cell acute lymphoblastic leukemia (B-ALL), the tumor that originates from pre-B cells, compared to common solid tumors and hematopoietic malignancies. While LGR5 is expressed in multiple solid tumors, including colon cancer,
Recommended publications
  • Mutant Luteinizing Hormone Receptors in a Compound Heterozygous Patient with Complete Leydig Cell Hypoplasia: Abnormal Processing Causes Signaling Deficiency
    0013-7227/02/$15.00/0 The Journal of Clinical Endocrinology & Metabolism 87(6):2506–2513 Printed in U.S.A. Copyright © 2002 by The Endocrine Society Mutant Luteinizing Hormone Receptors in a Compound Heterozygous Patient with Complete Leydig Cell Hypoplasia: Abnormal Processing Causes Signaling Deficiency J. W. M. MARTENS, S. LUMBROSO, M. VERHOEF-POST, V. GEORGET, A. RICHTER-UNRUH, M. SZARRAS-CZAPNIK, T. E. ROMER, H. G. BRUNNER, A. P. N. THEMMEN, AND CH. SULTAN Departments of Endocrinology and Reproduction and Internal Medicine, Erasmus University (J.W.M.M., M.V.-P., A.P.N.T.), 3000 DR Rotterdam, The Netherlands; Hormonologie du De´veloppement et de la Reproduction, Hoˆpital Lapeyronie and INSERM, U-439 (S.L., V.G., C.S.), 34090 Montpellier, France; Endocrinologie et Gyne´cologie Pe´diatriques, Hoˆpital A. de Villeneuve (C.S.), 34295 Montpellier, France; Department of Pediatric Endocrinology, University Children’s Hospital, University of Essen (A.R.-U.), 45122 Essen, Germany; Department of Pediatric Endocrinology, Children’s Memorial Health Institute (M.S.-C., T.E.R.), 04-730 Warsaw, Poland; and Department of Human Genetics, University Hospital (H.G.B.), 6500 HB Nijmegen, The Netherlands Over the past 5 yr several inactivating mutations in the absence of total and cell surface hormone binding, protein LH receptor gene have been demonstrated to cause Leydig levels of both mutant LH receptors are only moderately af- cell hypoplasia, a rare autosomal recessive form of male fected. The expression and study of enhanced green fluores- pseudohermaphroditism. Here, we report the identification of cent protein-tagged receptors confirmed this view and fur- two new LH receptor mutations in a compound heterozygous ther indicated that initial translocation to the endoplasmic case of complete Leydig hypoplasia and determine the cause reticulum of these mutant receptors is normal.
    [Show full text]
  • LGR5 and LGR6 in Stem Cell Biology and Ovarian Cancer
    www.impactjournals.com/oncotarget/ Oncotarget, 2018, Vol. 9, (No. 1), pp: 1346-1355 Review LGR5 and LGR6 in stem cell biology and ovarian cancer Adam J. Schindler1, Arisa Watanabe1 and Stephen B. Howell1 1Moores Cancer Center, University of California, San Diego, CA, USA Correspondence to: Stephen B. Howell, email: [email protected] Keywords: ovarian cancer, Wnt, LGR6, LGR5, RSPO Received: June 14, 2017 Accepted: July 31, 2017 Published: August 11, 2017 Copyright: Schindler et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License 3.0 (CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. ABSTRACT Wnt signaling plays a fundamental role in patterning of the embryo and maintenance of stem cells in numerous epithelia. Epithelial stem cells are closeted in niches created by surrounding differentiated cells that express secreted Wnt and R-spondin proteins that influence proliferation rate and fate determination of stem cell daughters. R-spondins act through the LGR receptors to enhance Wnt signaling. This close association of stem cells with more differentiated regulatory cells expressing Wnt-pathway ligands is a feature replicated in all of the epithelial stem cell systems thus far examined. How the stem cell niche operates through these short-range interactions is best understood for the crypts of the gastrointestinal epithelium and skin. Less well understood are the stem cells that function in the ovarian surface epithelium (OSE) and fallopian tube epithelium (FTE). While the cuboidal OSE appears to be made up of a single cell type, the cells of the FTE progress through a life cycle that involves differentiation into ciliated and secretory subtypes that are eventually shed into the lumen in a manner similar to the gastrointestinal epithelium.
    [Show full text]
  • Kynurenine Promotes the Goblet Cell Differentiation of HT-29 Colon Carcinoma Cells by Modulating Wnt, Notch and Ahr Signals
    1930 ONCOLOGY REPORTS 39: 1930-1938, 2018 Kynurenine promotes the goblet cell differentiation of HT-29 colon carcinoma cells by modulating Wnt, Notch and AhR signals JOO-HUNG PARK, JEONG-MIN LEE, EUN-JIN LEE, DA-JEONG KIM and WON-BHIN Hwang Department of Biology, Changwon National University, Changwon, Kyungnam 51140, Republic of Korea Received August 31, 2017; Accepted February 7, 2018 DOI: 10.3892/or.2018.6266 Abstract. Various amino acids regulate cell growth and inorganic salts for normal cell metabolism. However, the differentiation. In the present study, we examined the ability composition of medium formulations vary widely in concen- of HT-29 cells to differentiate into goblet cells in RPMI trations of glucose and other metabolic precursors, including and DMEM which are largely different in the amounts of amino acids. Thus, many types of cells showed different numerous amino acids. Most of the HT-29 cells differentiated responses to culture media in regards to proliferation and into goblet cells downregulating the stem cell marker Lgr5 differentiation. Th17 differentiation was induced more effi- when cultured in DMEM, but remained undifferentiated in ciently in Iscove's modified Dulbecco's medium (IMDM) than RPMI. The goblet cell differentiation in DMEM was inhibited in RPMI (1). Bone marrow-derived dendritic cells in IMDM by 1-methyl-tryptophan (1-MT), an inhibitor of indoleamine expressed higher levels of co-stimulatory and MHC II mole- 2,3 dioxygenase-1 which is the initial enzyme in tryptophan cules, compared to the cells generated in RPMI (2). In addition, metabolism along the kynurenine (KN) pathway, whereas hepatocyte differentiation and propagation and phenotype of tryptophan and KN induced goblet cell differentiation in corneal stroma-derived stem cells were modulated by culture RPMI.
    [Show full text]
  • Edinburgh Research Explorer
    Edinburgh Research Explorer International Union of Basic and Clinical Pharmacology. LXXXVIII. G protein-coupled receptor list Citation for published version: Davenport, AP, Alexander, SPH, Sharman, JL, Pawson, AJ, Benson, HE, Monaghan, AE, Liew, WC, Mpamhanga, CP, Bonner, TI, Neubig, RR, Pin, JP, Spedding, M & Harmar, AJ 2013, 'International Union of Basic and Clinical Pharmacology. LXXXVIII. G protein-coupled receptor list: recommendations for new pairings with cognate ligands', Pharmacological reviews, vol. 65, no. 3, pp. 967-86. https://doi.org/10.1124/pr.112.007179 Digital Object Identifier (DOI): 10.1124/pr.112.007179 Link: Link to publication record in Edinburgh Research Explorer Document Version: Publisher's PDF, also known as Version of record Published In: Pharmacological reviews Publisher Rights Statement: U.S. Government work not protected by U.S. copyright General rights Copyright for the publications made accessible via the Edinburgh Research Explorer is retained by the author(s) and / or other copyright owners and it is a condition of accessing these publications that users recognise and abide by the legal requirements associated with these rights. Take down policy The University of Edinburgh has made every reasonable effort to ensure that Edinburgh Research Explorer content complies with UK legislation. If you believe that the public display of this file breaches copyright please contact [email protected] providing details, and we will remove access to the work immediately and investigate your claim. Download date: 02. Oct. 2021 1521-0081/65/3/967–986$25.00 http://dx.doi.org/10.1124/pr.112.007179 PHARMACOLOGICAL REVIEWS Pharmacol Rev 65:967–986, July 2013 U.S.
    [Show full text]
  • Lgr5 Homologues Associate with Wnt Receptors and Mediate R-Spondin Signalling
    ARTICLE doi:10.1038/nature10337 Lgr5 homologues associate with Wnt receptors and mediate R-spondin signalling Wim de Lau1*, Nick Barker1{*, Teck Y. Low2, Bon-Kyoung Koo1, Vivian S. W. Li1, Hans Teunissen1, Pekka Kujala3, Andrea Haegebarth1{, Peter J. Peters3, Marc van de Wetering1, Daniel E. Stange1, Johan E. van Es1, Daniele Guardavaccaro1, Richard B. M. Schasfoort4, Yasuaki Mohri5, Katsuhiko Nishimori5, Shabaz Mohammed2, Albert J. R. Heck2 & Hans Clevers1 The adult stem cell marker Lgr5 and its relative Lgr4 are often co-expressed in Wnt-driven proliferative compartments. We find that conditional deletion of both genes in the mouse gut impairs Wnt target gene expression and results in the rapid demise of intestinal crypts, thus phenocopying Wnt pathway inhibition. Mass spectrometry demonstrates that Lgr4 and Lgr5 associate with the Frizzled/Lrp Wnt receptor complex. Each of the four R-spondins, secreted Wnt pathway agonists, can bind to Lgr4, -5 and -6. In HEK293 cells, RSPO1 enhances canonical WNT signals initiated by WNT3A. Removal of LGR4 does not affect WNT3A signalling, but abrogates the RSPO1-mediated signal enhancement, a phenomenon rescued by re-expression of LGR4, -5 or -6. Genetic deletion of Lgr4/5 in mouse intestinal crypt cultures phenocopies withdrawal of Rspo1 and can be rescued by Wnt pathway activation. Lgr5 homologues are facultative Wnt receptor components that mediate Wnt signal enhancement by soluble R-spondin proteins. These results will guide future studies towards the application of R-spondins for regenerative purposes of tissues expressing Lgr5 homologues. The genes Lgr4, Lgr5 and Lgr6 encode orphan 7-transmembrane 4–5 post-induction onwards.
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • A Benchmarking Study on Virtual Ligand Screening Against Homology Models of Human Gpcrs
    bioRxiv preprint doi: https://doi.org/10.1101/284075; this version posted March 19, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. A benchmarking study on virtual ligand screening against homology models of human GPCRs Victor Jun Yu Lima,b, Weina Dua, Yu Zong Chenc, Hao Fana,d aBioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, Matrix No. 07-01, 138671, Singapore; bSaw Swee Hock School of Public Health, National University of Singapore, 12 Science Drive 2, 117549, Singapore; cDepartment of Pharmacy, National University of Singapore, 18 Science Drive 4, 117543, Singapore; dDepartment of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore 117558 To whom correspondence should be addressed: Dr. Hao Fan, Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, Matrix No. 07-01, 138671, Singapore; Telephone: +65 64788500; Email: [email protected] Keywords: G-protein-coupled receptor, GPCR, X-ray crystallography, virtual screening, homology modelling, consensus enrichment 1 bioRxiv preprint doi: https://doi.org/10.1101/284075; this version posted March 19, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Abstract G-protein-coupled receptor (GPCR) is an important target class of proteins for drug discovery, with over 27% of FDA-approved drugs targeting GPCRs. However, being a membrane protein, it is difficult to obtain the 3D crystal structures of GPCRs for virtual screening of ligands by molecular docking.
    [Show full text]
  • Supplementary Table S5. Differentially Expressed Gene Lists of PD-1High CD39+ CD8 Tils According to 4-1BB Expression Compared to PD-1+ CD39- CD8 Tils
    BMJ Publishing Group Limited (BMJ) disclaims all liability and responsibility arising from any reliance Supplemental material placed on this supplemental material which has been supplied by the author(s) J Immunother Cancer Supplementary Table S5. Differentially expressed gene lists of PD-1high CD39+ CD8 TILs according to 4-1BB expression compared to PD-1+ CD39- CD8 TILs Up- or down- regulated genes in Up- or down- regulated genes Up- or down- regulated genes only PD-1high CD39+ CD8 TILs only in 4-1BBneg PD-1high CD39+ in 4-1BBpos PD-1high CD39+ CD8 compared to PD-1+ CD39- CD8 CD8 TILs compared to PD-1+ TILs compared to PD-1+ CD39- TILs CD39- CD8 TILs CD8 TILs IL7R KLRG1 TNFSF4 ENTPD1 DHRS3 LEF1 ITGA5 MKI67 PZP KLF3 RYR2 SIK1B ANK3 LYST PPP1R3B ETV1 ADAM28 H2AC13 CCR7 GFOD1 RASGRP2 ITGAX MAST4 RAD51AP1 MYO1E CLCF1 NEBL S1PR5 VCL MPP7 MS4A6A PHLDB1 GFPT2 TNF RPL3 SPRY4 VCAM1 B4GALT5 TIPARP TNS3 PDCD1 POLQ AKAP5 IL6ST LY9 PLXND1 PLEKHA1 NEU1 DGKH SPRY2 PLEKHG3 IKZF4 MTX3 PARK7 ATP8B4 SYT11 PTGER4 SORL1 RAB11FIP5 BRCA1 MAP4K3 NCR1 CCR4 S1PR1 PDE8A IFIT2 EPHA4 ARHGEF12 PAICS PELI2 LAT2 GPRASP1 TTN RPLP0 IL4I1 AUTS2 RPS3 CDCA3 NHS LONRF2 CDC42EP3 SLCO3A1 RRM2 ADAMTSL4 INPP5F ARHGAP31 ESCO2 ADRB2 CSF1 WDHD1 GOLIM4 CDK5RAP1 CD69 GLUL HJURP SHC4 GNLY TTC9 HELLS DPP4 IL23A PITPNC1 TOX ARHGEF9 EXO1 SLC4A4 CKAP4 CARMIL3 NHSL2 DZIP3 GINS1 FUT8 UBASH3B CDCA5 PDE7B SOGA1 CDC45 NR3C2 TRIB1 KIF14 TRAF5 LIMS1 PPP1R2C TNFRSF9 KLRC2 POLA1 CD80 ATP10D CDCA8 SETD7 IER2 PATL2 CCDC141 CD84 HSPA6 CYB561 MPHOSPH9 CLSPN KLRC1 PTMS SCML4 ZBTB10 CCL3 CA5B PIP5K1B WNT9A CCNH GEM IL18RAP GGH SARDH B3GNT7 C13orf46 SBF2 IKZF3 ZMAT1 TCF7 NECTIN1 H3C7 FOS PAG1 HECA SLC4A10 SLC35G2 PER1 P2RY1 NFKBIA WDR76 PLAUR KDM1A H1-5 TSHZ2 FAM102B HMMR GPR132 CCRL2 PARP8 A2M ST8SIA1 NUF2 IL5RA RBPMS UBE2T USP53 EEF1A1 PLAC8 LGR6 TMEM123 NEK2 SNAP47 PTGIS SH2B3 P2RY8 S100PBP PLEKHA7 CLNK CRIM1 MGAT5 YBX3 TP53INP1 DTL CFH FEZ1 MYB FRMD4B TSPAN5 STIL ITGA2 GOLGA6L10 MYBL2 AHI1 CAND2 GZMB RBPJ PELI1 HSPA1B KCNK5 GOLGA6L9 TICRR TPRG1 UBE2C AURKA Leem G, et al.
    [Show full text]
  • Mutational Landscape Differences Between Young-Onset and Older-Onset Breast Cancer Patients Nicole E
    Mealey et al. BMC Cancer (2020) 20:212 https://doi.org/10.1186/s12885-020-6684-z RESEARCH ARTICLE Open Access Mutational landscape differences between young-onset and older-onset breast cancer patients Nicole E. Mealey1 , Dylan E. O’Sullivan2 , Joy Pader3 , Yibing Ruan3 , Edwin Wang4 , May Lynn Quan1,5,6 and Darren R. Brenner1,3,5* Abstract Background: The incidence of breast cancer among young women (aged ≤40 years) has increased in North America and Europe. Fewer than 10% of cases among young women are attributable to inherited BRCA1 or BRCA2 mutations, suggesting an important role for somatic mutations. This study investigated genomic differences between young- and older-onset breast tumours. Methods: In this study we characterized the mutational landscape of 89 young-onset breast tumours (≤40 years) and examined differences with 949 older-onset tumours (> 40 years) using data from The Cancer Genome Atlas. We examined mutated genes, mutational load, and types of mutations. We used complementary R packages “deconstructSigs” and “SomaticSignatures” to extract mutational signatures. A recursively partitioned mixture model was used to identify whether combinations of mutational signatures were related to age of onset. Results: Older patients had a higher proportion of mutations in PIK3CA, CDH1, and MAP3K1 genes, while young- onset patients had a higher proportion of mutations in GATA3 and CTNNB1. Mutational load was lower for young- onset tumours, and a higher proportion of these mutations were C > A mutations, but a lower proportion were C > T mutations compared to older-onset tumours. The most common mutational signatures identified in both age groups were signatures 1 and 3 from the COSMIC database.
    [Show full text]
  • Cancer Stem Cells—Key Players in Tumor Relapse
    cancers Review Cancer Stem Cells—Key Players in Tumor Relapse Monica Marzagalli *, Fabrizio Fontana, Michela Raimondi and Patrizia Limonta Department of Pharmacological and Biomolecular Sciences, University of Milan, via Balzaretti 9, 20133 Milano, Italy; [email protected] (F.F.); [email protected] (M.R.); [email protected] (P.L.) * Correspondence: [email protected]; Tel.: +39-02-503-18427 Simple Summary: Cancer is one of the hardest pathologies to fight, being one of the main causes of death worldwide despite the constant development of novel therapeutic strategies. Therapeutic failure, followed by tumor relapse, might be explained by the existence of a subpopulation of cancer cells called cancer stem cells (CSCs). The survival advantage of CSCs relies on their ability to shape their phenotype against harmful conditions. This Review will summarize the molecular mechanisms exploited by CSCs in order to escape from different kind of therapies, shedding light on the potential novel CSC-specific targets for the development of innovative therapeutic approaches. Abstract: Tumor relapse and treatment failure are unfortunately common events for cancer patients, thus often rendering cancer an uncurable disease. Cancer stem cells (CSCs) are a subset of cancer cells endowed with tumor-initiating and self-renewal capacity, as well as with high adaptive abilities. Altogether, these features contribute to CSC survival after one or multiple therapeutic approaches, thus leading to treatment failure and tumor progression/relapse. Thus, elucidating the molecular mechanisms associated with stemness-driven resistance is crucial for the development of more effective drugs and durable responses. This review will highlight the mechanisms exploited by CSCs to overcome different therapeutic strategies, from chemo- and radiotherapies to targeted therapies Citation: Marzagalli, M.; Fontana, F.; and immunotherapies, shedding light on their plasticity as an insidious trait responsible for their Raimondi, M.; Limonta, P.
    [Show full text]
  • Unveiling Role of Sphingosine-1-Phosphate Receptor 2 As a Brake of Epithelial Stem Cell Proliferation and a Tumor Suppressor in Colorectal Cancer
    Unveiling role of Sphingosine-1-phosphate receptor 2 as a brake of epithelial stem cell proliferation and a tumor suppressor in colorectal cancer Luciana Petti Humanitas Clinical and Research Center-IRCCS Giulia Rizzo Humanitas University Federica Rubbino Humanitas University Sudharshan Elangovan Humanitas University Piergiuseppe Colombo Humanitas Clinical and Research Center-IRRCS Restelli Silvia Humanitas University Andrea Piontini Humanitas University Vincenzo Arena Policlinico Universitario Agostino Gemelli Michele Carvello Humanitas Clinical and Research Center-IRCCS Barbara Romano Universita degli Studi di Napoli Federico II Dipartimento di Medicina Clinica e Chirurgia Tommaso Cavalleri Humanitas Clinical and Research Center-IRCCS Achille Anselmo Humanitas Clinical and Research Center-IRCCS Federica Ungaro Humanitas University Silvia D’Alessio Humanitas University Antonino Spinelli Humanitas University Sanja Stifter Page 1/28 University of Rijeka Fabio Grizzi Humanitas Clinical and Research Center-IRCCS Alessandro Sgambato Istituto di Ricovero e Cura a Carattere Scientico Centro di Riferimento Oncologico della Basilicata Silvio Danese Humanitas University Luigi Laghi Universita degli Studi di Parma Alberto Malesci Humanitas University STEFANIA VETRANO ( [email protected] ) Humanitas University Research Keywords: colorectal cancer, Lgr5, S1PR2, PTEN, epithelial proliferation Posted Date: October 13th, 2020 DOI: https://doi.org/10.21203/rs.3.rs-56319/v2 License: This work is licensed under a Creative Commons Attribution 4.0 International License. Read Full License Version of Record: A version of this preprint was published on November 23rd, 2020. See the published version at https://doi.org/10.1186/s13046-020-01740-6. Page 2/28 Abstract Background. Sphingosine-1-phosphate receptor 2 (S1PR2) mediates pleiotropic functions encompassing cell proliferation, survival, and migration, which become collectively de-regulated in cancer.
    [Show full text]
  • G Protein-Coupled Receptors
    S.P.H. Alexander et al. The Concise Guide to PHARMACOLOGY 2015/16: G protein-coupled receptors. British Journal of Pharmacology (2015) 172, 5744–5869 THE CONCISE GUIDE TO PHARMACOLOGY 2015/16: G protein-coupled receptors Stephen PH Alexander1, Anthony P Davenport2, Eamonn Kelly3, Neil Marrion3, John A Peters4, Helen E Benson5, Elena Faccenda5, Adam J Pawson5, Joanna L Sharman5, Christopher Southan5, Jamie A Davies5 and CGTP Collaborators 1School of Biomedical Sciences, University of Nottingham Medical School, Nottingham, NG7 2UH, UK, 2Clinical Pharmacology Unit, University of Cambridge, Cambridge, CB2 0QQ, UK, 3School of Physiology and Pharmacology, University of Bristol, Bristol, BS8 1TD, UK, 4Neuroscience Division, Medical Education Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK, 5Centre for Integrative Physiology, University of Edinburgh, Edinburgh, EH8 9XD, UK Abstract The Concise Guide to PHARMACOLOGY 2015/16 provides concise overviews of the key properties of over 1750 human drug targets with their pharmacology, plus links to an open access knowledgebase of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties. The full contents can be found at http://onlinelibrary.wiley.com/doi/ 10.1111/bph.13348/full. G protein-coupled receptors are one of the eight major pharmacological targets into which the Guide is divided, with the others being: ligand-gated ion channels, voltage-gated ion channels, other ion channels, nuclear hormone receptors, catalytic receptors, enzymes and transporters. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading.
    [Show full text]